• Nenhum resultado encontrado

ADAMTS14 Gene Polymorphism and Environmental Risk in the Development of Oral Cancer.

N/A
N/A
Protected

Academic year: 2017

Share "ADAMTS14 Gene Polymorphism and Environmental Risk in the Development of Oral Cancer."

Copied!
11
0
0

Texto

(1)

ADAMTS14 Gene Polymorphism and

Environmental Risk in the Development of

Oral Cancer

Shih-Chi Su1,2, Ming-Ju Hsieh3,4,5, Yu-Fan Liu6,7, Ying-Erh Chou7,8, Chiao-Wen Lin9,10, Shun-Fa Yang3,7*

1Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan,2Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan,3Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan,4Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan,5Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan,6Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan,7Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan,8School of Medicine, Chung Shan Medical University, Taichung, Taiwan,9Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan,

10Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan

*ysf@csmu.edu.tw

Abstract

Background

Oral cancer is a common malignancy that is shown to be causally associated with heredi-tary and acquired factors. ADAMTS14 is a member of the ADAMTS (a disintegrin-like and metalloproteinase domain with thrombospondin motifs) metalloproteinase family that plays an important role in extracellular matrix (ECM) assembly and degradation. Elevation or defi-ciency of certain ADAMTS proteinases has been known to be implicated in a wide range of pathological processes including atherosclerosis, arthritis, and cancer. The present study aimed to explore the impact of ADAMTS14 gene polymorphisms, combined with environ-mental risks on the susceptibility to oral tumorigenesis.

Methodology/Principal Findings

Four single-nucleotide polymorphisms (SNPs) of the ADAMTS14 gene, including rs10823607, rs12774070, rs4747096, and rs61573157 were evaluated from 1200 normal controls and 850 patients with oral cancer. We failed to detect a significant association of four individual SNPs with oral cancer between case and control group. However, while considering behavioral exposure of environmental carcinogens, the presence of four ADAMTS14 SNPs, combined with betel nut chewing and/or smoking, profoundly leveraged the risk of oral cancer. Moreover, we observed a significant association of rs12774070, which is predicted to alter the expression and function of ADAMTS14 byin silicoand bioin-formatics analyses, with poor tumor cell differentiation (AOR: 0.59; 95% CI: 0.38–0.92;p= 0.02) in patients who chewed betel nuts.

a11111

OPEN ACCESS

Citation:Su S-C, Hsieh M-J, Liu Y-F, Chou Y-E, Lin C-W, Yang S-F (2016) ADAMTS14 Gene Polymorphism and Environmental Risk in the Development of Oral Cancer. PLoS ONE 11(7): e0159585. doi:10.1371/journal.pone.0159585

Editor:Amanda Ewart Toland, Ohio State University Medical Center, UNITED STATES

Received:April 6, 2016

Accepted:July 6, 2016

Published:July 27, 2016

Copyright:© 2016 Su et al. This is an open access article distributed under the terms of theCreative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Data Availability Statement:All relevant data are within the paper.

Funding:This work was supported in part by grants from the National Science Council, Taiwan (MOST 105-2628-B-040-003-MY3). This study was also supported by a grant from Chung Shan Medical University Hospital, Taiwan (CSH-2015-C-010). The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

(2)

Conclusions

These results implicate the interaction between ADAMTS14 gene polymorphisms and envi-ronmental mutagens as a risk factor of oral tumorigenesis and suggest a correlation of rs12774070 with the degree of oral tumor cell differentiation.

Introduction

Oral squamous cell carcinoma (OSCC) is a group of malignant lesions developing in the oral cav-ity and accounts for the vast majorcav-ity (approximately 90%) of oral neoplasms [1,2]. In spite of the current progresses in surgery and other treatment options, the fatality of oral cancer has remained mostly unchanged over the past decades [3,4]. The occurrence has not significantly improved although the etiological studies of oral cancer have uncovered several parameters that contribute to the development of OSCC [5–8]. It is demonstrated that recognized risk factors of oral cancer mainly include human papillomavirus (HPV) infection [5] and habitual exposure of cancer-causing substances, such as tobacco and alcohol use and betel nut chewing [6,9]. Besides, oral carcinogenesis is known to be modulated by genetic alterations that affect cell cycle, apopto-sis, and DNA repair [7,10]. Considering the heterogeneous nature and complex pathogenicity of cancer, all these factors seem to be reciprocal and needed to evaluate the disease prognosis.

ADAMTS (a disintegrin and metalloprotease with thrombospondin motifs) proteases repre-sent an extracellular zinc metalloproteinase family with 19 members in humans. These multido-main proteases are known to catalyze a great variety of substrates in the extracellular matrices (ECM) [11]. Current studies have unveiled their association with a wide range of physiological and pathological processes including blood coagulation, atherosclerosis, wound healing, angio-genesis, fertility, arthritis and cancer [12]. ADAMTS14 is a recently discovered procollagen N-propeptidase that functions in reducing the solubility of the collagen molecules and facilitating their assembly into cylindrical collagen fibrils. In addition to its enzymatic activity, ADAMTS14, sharing a sequence and functional homology with ADAMTS2 and ADAMTS3, is characterized by the presence of four thrombospondin type I domains (TSR1) and a C-terminal procollagen N-proteinase (PNP) domain comprising a“protease and lacunin”(PLAC) domain. An anti-tumor effect of ADAMTS2 through interrupting intraanti-tumoral vascularization has been demon-strated [13]. Although at a lower level, ADAMTS14 is usually co-expressed with ADAMTS2 in all type I collagen-rich connective tissues to be responsible for ECM remodeling [14].

Genetic associations of ADAMTS14 with several conditions have been documented. Poly-morphisms within the ADAMTS14 gene have been shown to influence genetic predisposition to multiple sclerosis [15]. Several other studies have implicated ADAMTS14 gene variations as a risk component in knee osteoarthritis and Achilles tendon pathology [16–18]. In recent years, there has been an explosion of reports concerning how these ADAMTS proteases can influence tumor microenvironment to potentiate cancer progression. However, little is known regarding the joint effects of ADAMTS14 gene polymorphisms and behavioral exposure of cancer-causing substances on the predisposition to oral cancer. Here, we conducted a hypothe-sis-driven case-control study to explore the effects of the interactions of ADAMTS14 gene polymorphisms with the environmental carcinogens on the risk of OSCC.

Materials and Methods

Subjects

(3)

Chung Shan Medical University Hospital in Taichung, Taiwan. Participants were recruited from 2008 to 2015. Among the 850 cases, tumors were located in the buccal mucosa, tongue, gingiva, palate, floor of the mouth, and others. Oral cancer patients were staged clinically at the time of diagnosis according to the TNM staging system of the American Joint Committee on Cancer (AJCC) [19]. Tumor differentiation was examined by a pathologist and rated according to the AJCC classification. Subjects with neither self-reported history of cancer of any sites nor oral precancerous disease such as oral submucous fibrosis, leukoplakia, erythroplakia, verru-cous hyperplasia, etc. were recruited to the control group. All participants provided informed written consent at enrollment. Data on age, gender, alcohol drinking, betel quid chewing, and cigarette smoking were recorded from each participant. Betel quid chewing and alcohol drink-ing are defined as behavioral use of betel nuts (or related products) and alcoholic drinkdrink-ing, respectively. Cigarette smoking is defined as current smoking of at least one cigarette per day during the latest three months. The Institutional Review Board of Chung Shan Medical Univer-sity Hospital approved this study (CSMUH No: CS13214-1), and informed written consent was obtained from each participant.

Selection and genotyping of SNPs

In the present study, the selection of four common polymorphisms fromADAMTS14gene is based on their wide associations with the development of miscellaneous pathophysiological processes [16,18,20,21]. To obtain adequate power for testing the potential association and evaluate the putative functional relevance of ADAMTS14, four non-synonymous SNPs, includ-ing rs10823607, rs12774070, rs4747096, and rs61573157, with minor allele frequencies>5%

were chosen (Table 1). Genomic DNA was isolated from 3 mL of blood using QIAamp DNA blood mini kits (Qiagen, Valencia, CA, USA). Assessment of allelic discrimination for the

ADAMTS14SNPs was performed by using the TaqMan assay with an ABI StepOne™

Real-Time PCR System (Applied Biosystems, Foster City, CA, USA), and further evaluated with SDS version 3.0 software (Applied Biosystems). The total volume of TaqMan assays was 10μL,

containing 5μL of Master Mix, 0.25μL of probes, and 10 ng of genomic DNA. The real-time

PCR reaction included an initial denaturation step at 95°C for 10 min, followed by 40 amplifi-cation cycles, each consisting of 95°C for 15 sec and 60°C for 1 min.

Statistical analysis

A chi-square goodness-of-fit test was used to evaluate Hardy-Weinberg equilibrium for bialle-lic markers. The differences in demographic parameters between OSCC patients and

cancer-Table 1. ADAMTS14gene polymorphisms assessed in this study.

Variable Exon (contiguous position)

Chromosome 10:70741007 10:70753879 10:70758253 10:70760503

Exon 12 19 21 22

cDNA position & nucleotide change╪ c.1778T>C c.2818C>A c.3155A>G c.3331C>T Protein position†

593 940 1052 1111

dbSNP rs No. rs10823607 rs12774070 rs4747096 rs61573157

Function Nonsynonymous Nonsynonymous Nonsynonymous Nonsynonymous

dbSNP allele CTG>CGG CTG>ATG GAA>GGA CCA>TCA

Protein residue L [Leu]>P [Pro] L [Leu]>M [Met] E [Glu]>G [Gly] P [Pro]>S [Ser]

Codon position 2 1 2 1

╪: NM_139155.2 †: NP_631894.2.

(4)

free controls were estimated by using Fisher’s exact test or parametric independent t-test. The adjusted odds ratios (AORs) with their 95% confidence intervals (CIs) for the association between genotype frequencies and the risk of OSCC plus clinicopathological characteristics were estimated by multiple logistic regression models after controlling for other covariates. The interactions of ADAMTS14 gene polymorphisms with each demographic characteristic were evaluated as described previously [22]. The association of rs12774070 genotypes with the expression levels of ADAMTS14 in esophagus mucosa tissues was assessed by one-way ANOVA. Apvalue<0.05 was considered significant. The data were analyzed by using SAS

statistical software (Version 9.1, 2005; SAS Institute Inc., Cary, NC).

Bioinformatics analysis

Several bioinformatics tools were used to assess a putative functional relevance of rs12774070. Data from the Genotype-Tissue Expression (GTEx) database were used to identify the correla-tions between rs12774070 and ADAMTS14 expression in esophagus mucosa tissues [23]. The homology model of TSR1 (3) domain of ADAMTS14 was illustrated with the ribbon diagram (ViewerLite 5.0) by using Swiss Model sever (template as PDB accession number: 3GHM, 40.43% identity). Candidate deleterious non-synonymous SNPs were identified using SIFT [24] and PolyPhen-2 [25].

Results

In this study, 850 patients with oral cancer and 1200 normal controls were recruited to explore the risk effect of ADAMTS14 gene polymorphisms (rs10823607, rs12774070, rs4747096, and rs61573157) on the development of oral neoplasm. The demographic characteristics of two study groups as well as the clinical parameters of cancer patients were evaluated (Table 2). Only male participants are enrolled to bypass the impact of gender differences on disease etiol-ogy. In accordance with our previous data [22] and findings from others [3,4], significant dif-ferences in smoking, alcohol drinking, and betel nut chewing were observed between healthy participants and OSCC patients.

To test the potential influence of ADAMTS14 gene polymorphisms on the development of OSCC, four non-synonymous single-nucleotide polymorphisms (nsSNPs), rs10823607, rs12774070, rs4747096, and rs61573157 were evaluated in this investigation. The distributions of genotype frequencies for each SNP were shown inTable 3. In these controls, the genotypic frequency of ADAMTS14 SNP rs10823607, rs12774070, rs4747096, and rs61573157 met the Hardy-Weinberg equilibrium (p= 0.87,χ2value: 0.02;p= 0.10,χ2value: 2.76;p= 0.26,χ2

value: 1.27; andp= 0.65,χ2value: 0.20, respectively). Moreover, in the case group, the

frequen-cies of four selected SNPs also met the Hardy-Weinberg equilibrium (p = 0.42,χ2 value: 0.64;

p = 0.37,χ2 value: 0.79; p = 0.82,χ2 value: 0.05; and p = 0.98,χ2 value:<0.01, respectively).

(5)

16.72; 95%CI, 11.80–23.69) (Table 4). This risk is further heightened when individuals who not only chew betel nut but also carry at least one minor allele of rs10823607, rs12774070,

rs4747096, and rs61573157 (rs10823607: AOR, 14.45; 95%CI, 8.38–24.93; rs12774070: AOR, 15.36; 95%CI, 9.53–24.77; rs4747096: AOR, 17.17; 95%CI, 10.55–27.94; and rs61573157: AOR, 13.90; 95%CI, 8.13–23.76). Though the genetic contribution alone appears to be subtle, a signif-icant interaction between betel nut chewing and the existence of at least one polymorphic allele of these four ADAMTS14 SNPs is shown to be correlated with the incidence of oral cancer in smokers.

We further analyzed the combined effect of ADAMTS14 gene polymorphisms and environ-mental factors on influencing the clinical status of OSCC. For 694 patients who chewed betel quid, a significant association of rs12774070 variants (CA+AA vs CC) with poor tumor cell dif-ferentiation (AOR: 0.62; 95% CI: 0.41–0.94;p= 0.02) but not clinical stage, tumor size, and lymph node metastasis was observed (Table 5). However, we failed to detect any significant asso-ciation of the other 3 ADAMTS14 variants individually with the clinical status of OSCC in betel nut users (Data not shown). Moreover, as a preliminary assessment of the putative functional rel-evance of ADAMTS14 rs12774070, alteration in ADAMTS14 expression was seen in esophagus mucosa tissues of individuals who carry polymorphic allele of ADAMTS14 rs12774070 in the

Table 2. The distributions of demographical characteristics in 1200 male controls and 850 male patients with oral cancer.

Variable Controls (N = 1200) Patients (N = 850) pvalue

Age (yrs) Mean±S.D. Mean±S.D.

53.91±10.02 54.60±11.09 p= 0.14

Betel quid chewing

No 1001 (83.4%) 156 (18.4%)

Yes 199 (16.6%) 694 (81.6%) p<0.01*

Cigarette smoking

No 564 (47.0%) 85 (10.0%)

Yes 636 (53.0%) 765 (90.0%) p<0.01*

Alcohol drinking

No 963 (80.3%) 357 (42.0%)

Yes 237 (19.7%) 493 (58.0%) p<0.01*

Stage

I+II 412 (48.5%)

III+IV 438 (51.5%)

Tumor T status

T1+T2 489 (57.5%)

T3+T4 361 (42.5%)

Lymph node status

N0 570 (67.1%)

N1+N2+N3 280 (32.9%)

Metastasis

M0 841 (98.9%)

M1 9 (1.1%)

Cell differentiation

Well differentiated 141 (16.6%)

Moderately or poorly differentiated 709 (83.4%)

Parametric independent t-test was used between healthy controls and patients with oral cancer. *pvalue<0.05 as statistically significant.

(6)

Genotype-Tissue Expression (GTEx) database (Fig 1A). The change in protein sequence due to the polymorphic allele of ADAMTS14 rs12774070 resides in a region of TSR1 domain, which is conserved among procollagen N-propeptidases (Fig 1B–1D). Furthermore, computational algo-rithms that are widely used for the discrimination of damaging nsSNPs from neutral ones have predicted ADAMTS14 rs12774070 to be deleterious (Fig 1E). There data suggest that changes in ADAMTS14 expression and function due to genetic polymorphisms, in combination with betel quid chewing, may affect tumor cell differentiation of oral cancer.

Discussion

Converging lines of evidence has supported that development of OSCC is a complicated pro-cess regulated by both environmental and genetic factors. In the present study, we for the first time revealed that the combination of the ADAMTS14 gene polymorphisms with the behav-ioral use of betel nut and tobacco conferred a higher susceptibility to oral cancer. Strikingly, betel nut users bearing the polymorphic allele of ADAMTS14 rs12774070 were shown to be less likely to develop tumors with moderate/poor differentiation.

The ADAMTS family has been examined intensively for its proteolytic function on tumor angiogenesis, an indispensable process vital for cancer progression and metastasis [26]. Para-doxically, the activation of ADAMTS proteinases can exhibit both inhibitory and promotive effects on angiogenesis as the mechanism involved in their regulation of cancer development varies among different members [27]. Some members (ADAMTS2, 4, 6, and 14) are shown to be manifestly upregulated in cancer tissues, whereas others (ADAMTS1, 5, 8, 9, 10, 13, 15, and

Table 3. Distributions of genotypic frequencies ofADAMTS14SNPs in controls and oral cancer patients.

Variable Controls (N = 1200) n (%) Patients (N = 850) n (%) OR (95% CI) AOR (95% CI)

rs10823607

CC 1017 (84.7%) 714 (84.0%) 1.00 1.00

CT 175 (14.6%) 132 (15.5%) 1.07 (0.84–1.37) 1.03 (0.74–1.42)

TT 8 (0.7%) 4 (0.5%) 0.71 (0.21–2.37) 0.80 (0.15–4.18)

CT+TT 183 (15.3%) 136 (16.0%) 1.06 (0.83–1.35) 1.02 (0.74–1.40)

rs12774070

CC 876 (73.0%) 616 (72.5%) 1.00 1.00

CA 290 (24.2%) 219 (25.8%) 1.07 (0.88–1.32) 1.06 (0.81–1.39)

AA 34 (2.8%) 15 (1.8%) 0.63 (0.34–1.16) 0.47 (0.21–1.07)

CA+AA 324 (27.0%) 234 (27.5%) 1.03 (0.84–1.25) 0.99 (0.76–1.29)

rs4747096

AA 489 (40.8%) 360 (42.4%) 1.00 1.00

AG 568 (47.3%) 384 (45.2%) 0.92 (0.76–1.11) 1.00 (0.78–1.29)

GG 143 (12.0%) 106 (12.5%) 1.01 (0.76–1.34) 1.19 (0.81–1.75)

AG+GG 711 (59.3%) 490 (57.6%) 0.94 (0.78–1.12) 1.04 (0.82–1.32)

rs61573157

CC 995 (82.9%) 702 (82.6%) 1.00 1.00

CT 194 (16.2%) 141 (16.6%) 1.03 (0.81–1.31) 1.03 (0.75–1.41)

TT 11 (0.9%) 7 (0.8%) 0.90 (0.35–2.34) 1.31 (0.36–4.85)

CT+TT 205 (17.1%) 148 (17.4%) 1.02 (0.81–1.29) 1.04 (0.76–1.42)

The odds ratio (OR) with their 95% confidence intervals were estimated by logistic regression models.

The adjusted odds ratio (AOR) with their 95% confidence intervals were estimated by multiple logistic regression models after controlling for betel nut chewing, alcohol and tobacco consumption.

(7)

18) are downregulated [28,29]. In the present study, we showed a significant interaction between betel nut chewing and the presence of at least one polymorphic allele of four ADAMTS14 SNPs examined is associated with higher incidence of oral cancer in smokers. In addition, a significant increase in ADAMTS14 expression was observed in esophagus mucosa tissues of subjects who possess the polymorphic allele of ADAMTS14 rs12774070 (Fig 1A). Unlike the ADAMs (a disintegrin and metalloproteinases), the ADAMTS proteinases are secreted by the cells and are not membrane-bound. Since oral cancer is a type of malignancies that develops in the mucosa tissues of the mouth or throat, it is conceivable that the accumula-tion of carcinogenic substances derived from addictive behaviors (e.g. betel nut chewing and smoking) over time in combination with the elevation of specific regulators of tumor angiogen-esis, such as ADAMTS14, within oral or adjacent tissues could finally lead to a malignant phenotype.

More intriguingly, we identified an inverse association of ADAMTS14 rs12774070 with can-cer cell differentiation in the present study. Due to a relatively small sample size, further vali-dating this observation will require additional experimental evidence in future investigations. The degree of epithelial cell differentiation in oral cancer is determined by the level of keratini-zation, a process that hardened cells filled with structurally and functionally distinct keratin [30]. A lack of cell differentiation in OSCC has been associated with more rapid tumor growth

Table 4. Associations of combined effects ofADAMTS14genotypic frequencies and betel nut chewing with oral cancer among 1401 smokers.

Variable Controls (n = 636) (%) Patients (n = 765) (%) OR (95% CI) AOR (95% CI)

rs10823607

aCC genotype & non-betel nut chewing 372 (58.5%) 91 (11.9%) 1.00 1.00

bCT or TT genotype & non-betel nut chewing 77 (12.1%) 13 (1.7%) 0.70 (0.37–1.30) 0.54 (0.26–1.13) c

CC genotype with betel nut chewing 160 (25.2) 556 (72.7%) 14.21 (10.64–18.97) 13.07 (9.40–18.18) dCT or TT genotype with betel nut chewing 27 (4.2%) 105 (13.7%) 15.90 (9.83

–25.72) 14.45 (8.38–24.93) rs12774070

aCC genotype & non-betel nut chewing 326 (51.3%) 75 (9.8%) 1.00 1.00

b

CA or AA genotype & non-betel nut chewing 123 (19.3%) 29 (3.8%) 1.03 (0.64–1.65) 0.89 (0.51–1.55) cCC genotype with betel nut chewing 137 (21.5%) 478 (62.5%) 15.17 (11.07

–20.78) 15.05 (10.43–21.70) dCA or AA genotype with betel nut chewing 50 (7.9%) 183 (23.9%) 15.91 (10.65

–23.76) 15.36 (9.53–24.77) rs4747096

a

AA genotype & non-betel nut chewing 190 (29.9%) 36 (4.7%) 1.00 1.00

bAG or GG genotype & non-betel nut chewing 259 (40.7%) 68 (8.9%) 1.39 (0.89–2.16) 1.33 (0.80–2.22) cAA genotype with betel nut chewing 73 (11.5%) 293 (38.3%) 21.18 (13.66

–32.86) 24.74 (14.38–42.57) dAG or GG genotype with betel nut chewing 114 (17.9%) 368 (48.1%) 17.04 (11.27

–25.76) 17.17 (10.55–27.94) rs61573157

aCC non-betel nut chewing 375 (59.0%) 79 (10.3%) 1.00 1.00

bCT or TT genotype & non-betel nut chewing 74 (11.6%) 25 (3.3%) 1.60 (0.96–2.68) 1.58 (0.86–2.89) cCC genotype with betel nut chewing 152 (23.9%) 549 (71.8%) 17.15 (12.68

–23.19) 16.72 (11.80–23.69) c

CT or TT genotype with betel nut chewing 35 (5.5%) 112 (14.6%) 15.19 (9.68–23.83) 13.90 (8.13–23.76)

The odds ratios (ORs) with their 95% confidence intervals were estimated by logistic regression models.

The adjusted odds ratios (AORs) with their 95% confidence intervals were estimated by multiple logistic regression models after controlling for age and alcohol consumption.

a

Individual with wild-type genotype but without betel nut chewing. bIndividual with either at least one minor allele without betel nut chewing. c

Individual with wild-type genotype and betel nut chewing. d

Individual with both at least one minor allele and betel nut chewing.

(8)

and spread, while specific keratin expression patterns are used as an independent prognostic marker and indicate a decreased overall and progression-free survival [31]. Several factors, such as components of ECM, growth factors (e.g. epidermal growth factor, transforming growth factor alpha and beta), retinoids and calcium, are known to regulate epithelial differen-tiation [30,32]. ADAMTS proteinases catalyze a great variety of ECM molecules, and the ECM also controls the activity and presentation of a wide range of growth factors [33]. Therefore, remodeling of the ECM by ADAMTSs has profound effects on cell differentiation and the con-sequent clinical manifestations of oral cancer. Moreover, rs12774070-derived alteration in pro-tein sequence resides in a conserved region of TSR1(3) domain and adjacent to a bisulfide bridge and N-linked glycosylation site (Fig 1). Predictions from sequence- and structure-based algorithms show that the presence of polymorphic allele of ADAMTS14 rs12774070, rather than rs10823607, rs4747096, and rs61573157, confers a determinant impact on ADAMTS14 function. These data indicate that altered ADAMTS14 function due to genetic polymorphisms, in combination with betel quid chewing, may affect tumor cell differentiation of oral cancer by virtue of disturbance of ECM remodeling.

We presented an impact of AMAMTS14 gene variations in combination with acquired fac-tors on the incidence and tumor cell differentiation of OSCC; however, there are several limita-tions in the study. One is that all habitual exposures of carcinogens were dichotomized. The effects of environmental risks on the predisposition to oral cancer may be underestimtated because of a lack of quantitative definition for betel nut chewing, drinking, and smoking. The other concern is that esophageal mucosa was selected based on anatomical relevance for evalu-ation from GTEx database because oral tissue is unavailable. Also unavailable are patients’ sur-vival data since a majority of patients were enrolled in the past few years. Another weakness is that the control group in this hospital-based study was recruited among individuals without cancer. We could not entirely exclude the possibility of potential selection and recall bias. In addition, the results presented in this study may not be able to be extended to other popula-tions unless replication studies are conducted. Furthermore, the functional role of ADAMTS14 in cell differentiation of oral cancer requires further investigation.

Table 5. Associations of genotype frequencies ofADAMTS14rs12774070 with clinical status of oral cancer among 694 betel quid users.

Variable ADAMTS14rs12774070 (betel quid users)

CC (n = 503) n (%) CA+AA (n = 191) n (%) OR (95% CI)pvalue AOR (95% CI)pvalue

Clinical Stage

Stage I/II 248 (49.3%) 91 (47.6%) 1.00 1.00

Stage III/IV 255 (50.7%) 100 (52.4%) 1.07 (0.77–1.49)p= 0.70 1.10 (0.77–1.58)p= 0.60

Tumor size

T2 288 (57.3%) 109 (57.1%) 1.00 1.00

>T2 215 (42.7%) 82 (42.9%) 1.01 (0.72–1.41)p= 0.96 1.07 (0.75–1.54)p= 0.71

Lymph node metastasis

No 339 (67.4%) 127 (66.5%) 1.00 1.00

Yes 164 (32.6%) 64 (33.5%) 1.04 (0.73–1.45)p= 0.82 1.08 (0.73–1.56)p= 0.72

Cell differentiation

well 79 (15.7%) 44 (23.0%) 1.00 1.00

Moderate/poor 424 (84.3%) 147 (77.0%) 0.62 (0.41–0.94)p= 0.02* 0.59 (0.38–0.92)p= 0.02*

The adjusted odds ratios (AORs) with their 95% confidence intervals were estimated by multiple logistic regression models after controlling for age, alcohol and tobacco consumption.

*p<0.05.

(9)

Fig 1. Functional implication andIn silicoprofiling of ADAMTS14 SNP rs12774070. (A)ADAMTS14 displays a significant eQTL association with rs12774070 genotypes in esophagus mucosa tissues (GTEx data set). (B) The altered residue is conserved throughout three procollagen aminopropeptidase subfamily of ADAMTS proteases, including ADAMTS2 (NP_055059.2), ADAMTS3 (NP_055058.2) and ADAMTS14 (NP_631894.2), as shown by alignment of the protein sequences with Crustal Omega software. Numbering is for human ADMDTS14. The arrow represents b-strands, and consensus sites for N-liked oligosaccharide attachment are marked in red (pinpointed). The disulfide bonds are marked in dark lines connecting paired cysteines. (C) Schematic representation of the full-length human ADMDTS14 protein, domain symbols are drawn approximately to scale. The rectangles represent the key domain structures, ZnMc superfamily (cd04273; pink), ADAM spacer1 (pfam05986; green) and four TSR1 (smart00209; yellow) by NCBI CDD server. Putative furin cleavage site is indicated by an arrowhead, and N-linked glycosylation sites are pointed by pins. (D) Ribbon diagram depicts the homology model of TSR1 (3) domain of ADAMTS14. The ribbon indicates the Cαcarbon of the TSR1 domain characterized in this study. The blue ribbon, purple sphere,

green spheres and yellow sticks indicate the b-strands structure, SNP rs12774070, potential N-linked glycosylation site and disulfide bonds, respectively. (E) Pathogenicity prediction of ADAMTS14 SNPs assessed in this study.

(10)

In conclusion, our results demonstrate that a joint effect of ADAMTS14 SNPs (rs10823607, rs12774070, rs4747096, and rs61573157) with betel nut chewing and smoking causally contrib-utes to the occurrence of oral cancer. In addition, an inverse association of SNP ADAMTS14 rs12774070 with tumors cell differentiation was observed in betel nut users. These findings reveal a novel genetic-environmental predisposition to oral tumorigenesis.

Acknowledgments

This work was supported in part by grants from the National Science Council, Taiwan (MOST 105-2628-B-040-003-MY3). This study was also supported by a grant from Chung Shan Medi-cal University Hospital, Taiwan (CSH-2015-C-010). The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Author Contributions

Conceived and designed the experiments: SCS SFY. Performed the experiments: YFL MJH YEC. Analyzed the data: MJH CWL. Contributed reagents/materials/analysis tools: SFY. Wrote the paper: SCS YFL SFY.

References

1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, et al. Cancer statistics, 2008. CA Cancer J Clin 2008; 58: 71–96. doi:10.3322/CA.2007.0010PMID:18287387

2. Krishna Rao SV, Mejia G, Roberts-Thomson K and Logan R. Epidemiology of oral cancer in Asia in the past decade—an update (2000–2012). Asian Pac J Cancer Prev 2013; 14: 5567–5577. PMID: 24289546

3. Jadhav KB and Gupta N. Clinicopathological Prognostic Implicators of Oral Squamous Cell Carcinoma: Need to Understand and Revise. N Am J Med Sci 2013; 5: 671–679. doi:10.4103/1947-2714.123239 PMID:24404549

4. Zini A, Czerninski R and Sgan-Cohen HD. Oral cancer over four decades: epidemiology, trends, histol-ogy, and survival by anatomical sites. J Oral Pathol Med 2010; 39: 299–305. doi:10.1111/j.1600-0714. 2009.00845.xPMID:20040019

5. Gupta K and Metgud R. Evidences Suggesting Involvement of Viruses in Oral Squamous Cell Carci-noma. Patholog Res Int 2013; 2013: 642496. doi:10.1155/2013/642496PMID:24455418

6. Scully C and Bagan J. Oral squamous cell carcinoma overview. Oral Oncol 2009; 45: 301–308. doi: 10.1016/j.oraloncology.2009.01.004PMID:19249237

7. Scully C, Field JK and Tanzawa H. Genetic aberrations in oral or head and neck squamous cell carci-noma (SCCHN): 1. Carcinogen metabolism, DNA repair and cell cycle control. Oral Oncol 2000; 36: 256–263. PMID:10793327

8. Scully C, Field JK and Tanzawa H. Genetic aberrations in oral or head and neck squamous cell carci-noma 2: chromosomal aberrations. Oral Oncol 2000; 36: 311–327. PMID:10899669

9. Awang MN. Betel quid and oral carcinogenesis. Singapore Med J 1988; 29: 589–593. PMID:3075344

10. Taniyama Y, Takeuchi S and Kuroda Y. Genetic polymorphisms and oral cancer. J UOEH 2010; 32: 221–236. PMID:20857816

11. Apte SS. A disintegrin-like and metalloprotease (reprolysin-type) with thrombospondin type 1 motif (ADAMTS) superfamily: functions and mechanisms. J Biol Chem 2009; 284: 31493–31497. doi:10. 1074/jbc.R109.052340PMID:19734141

12. Le Goff C and Cormier-Daire V. The ADAMTS(L) family and human genetic disorders. Hum Mol Genet 2011; 20: R163–167. doi:10.1093/hmg/ddr361PMID:21880666

13. Dubail J, Kesteloot F, Deroanne C, Motte P, Lambert V, Rakic JM, et al. ADAMTS-2 functions as anti-angiogenic and anti-tumoral molecule independently of its catalytic activity. Cell Mol Life Sci 2010; 67: 4213–4232. doi:10.1007/s00018-010-0431-6PMID:20574651

14. Bekhouche M and Colige A. The procollagen N-proteinases ADAMTS2, 3 and 14 in pathophysiology. Matrix Biol 2015; 44–46: 46–53. doi:10.1016/j.matbio.2015.04.001PMID:25863161

(11)

16. El Khoury L, Posthumus M, Collins M, Handley CJ, Cook J and Raleigh SM. Polymorphic variation within the ADAMTS2, ADAMTS14, ADAMTS5, ADAM12 and TIMP2 genes and the risk of Achilles ten-don pathology: a genetic association study. J Sci Med Sport 2013; 16: 493–498. doi:10.1016/j.jsams. 2013.02.006PMID:23491141

17. Poonpet T, Honsawek S, Tammachote N, Kanitnate S and Tammachote R. ADAMTS14 gene polymor-phism associated with knee osteoarthritis in Thai women. Genet Mol Res 2013; 12: 5301–5309. doi: 10.4238/2013.November.7.5PMID:24301791

18. Rodriguez-Lopez J, Pombo-Suarez M, Loughlin J, Tsezou A, Blanco FJ, Meulenbelt I, et al. Association of a nsSNP in ADAMTS14 to some osteoarthritis phenotypes. Osteoarthritis Cartilage 2009; 17: 321– 327. doi:10.1016/j.joca.2008.07.012PMID:18790654

19. Edge SB and Compton CC. The American Joint Committee on Cancer: the 7th edition of the AJCC can-cer staging manual and the future of TNM. Ann Surg Oncol 2010; 17: 1471–1474. doi:10.1245/ s10434-010-0985-4PMID:20180029

20. Byrne EM, Gehrman PR, Medland SE, Nyholt DR, Heath AC, Madden PA, Hickie IB, Van Duijn CM, Henders AK, Montgomery GW, Martin NG, Wray NR and Chronogen C. A genome-wide association study of sleep habits and insomnia. Am J Med Genet B Neuropsychiatr Genet 2013; 162B: 439–451. doi:10.1002/ajmg.b.32168PMID:23728906

21. Grupe A, Li Y, Rowland C, Nowotny P, Hinrichs AL, Smemo S, et al. A scan of chromosome 10 identi-fies a novel locus showing strong association with late-onset Alzheimer disease. Am J Hum Genet 2006; 78: 78–88. PMID:16385451

22. Su S, Chien M, Lin C, Chen M and Yang S. RAGE gene polymorphism and environmental factor in the risk of oral cancer. J Dent Res 2015; 94: 403–411. doi:10.1177/0022034514566215PMID:25582438

23. Consortium GT. The Genotype-Tissue Expression (GTEx) project. Nat Genet 2013; 45: 580–585. doi: 10.1038/ng.2653PMID:23715323

24. Ng PC and Henikoff S. Predicting deleterious amino acid substitutions. Genome Res 2001; 11: 863– 874. PMID:11337480

25. Adzhubei I, Jordan DM and Sunyaev SR. Predicting functional effect of human missense mutations using PolyPhen-2. Curr Protoc Hum Genet 2013; Chapter 7: Unit7 20.

26. Sun Y, Huang J and Yang Z. The roles of ADAMTS in angiogenesis and cancer. Tumour Biol 2015; 36: 4039–4051. doi:10.1007/s13277-015-3461-8PMID:25916206

27. Cal S and Lopez-Otin C. ADAMTS proteases and cancer. Matrix Biol 2015; 44–46: 77–85. doi:10. 1016/j.matbio.2015.01.013PMID:25636539

28. Kumar S, Rao N and Ge R. Emerging Roles of ADAMTSs in Angiogenesis and Cancer. Cancers (Basel) 2012; 4: 1252–1299.

29. Wagstaff L, Kelwick R, Decock J and Edwards DR. The roles of ADAMTS metalloproteinases in tumori-genesis and metastasis. Front Biosci (Landmark Ed) 2011; 16: 1861–1872.

30. Shetty S and Gokul S. Keratinization and its disorders. Oman Med J 2012; 27: 348–357. doi:10.5001/ omj.2012.90PMID:23074543

31. Fillies T, Werkmeister R, Packeisen J, Brandt B, Morin P, Weingart D, Joos U and Buerger H. Cytokera-tin 8/18 expression indicates a poor prognosis in squamous cell carcinomas of the oral cavity. BMC Cancer 2006; 6: 10. PMID:16412231

32. Squier CA and Kremer MJ. Biology of oral mucosa and esophagus. J Natl Cancer Inst Monogr 2001; 7– 15. PMID:11694559

Referências

Documentos relacionados

Com relação à análise da capacidade de uma Escola Metropolitana de Administração Pública em oportunizar ao servidor uma formação adequada, compreendendo a

In the present study, the association between CYP1A1 gene polymorphisms (IVS1-728G &gt; A, Thr461Asn and Ile462Val), and the risk of oral cancer, was examined among 157 patients

The genetic alterations observed in head and neck cancer are mainly due to oncogene activation (gain of function mutations) and tumor suppressor gene inactivation (loss of

Primarily, the tumor suppressor gene expression was assigned to genes involved in the control of strategic points of the chain of events related to cell growth and

Estrutura para análises fisico-químicas laboratoriais 23 Pesca com 'galão' realizada no açude Paus Branco, Madalena/CE 28 Macrófitas aquáticas flutuantes no nude Paus

M ethylenetetrahydrofolate reductase gene (MTHFR) C677T polymorphism may be a risk factor for head and neck squamous cell carcinoma due to changes in folate levels that can

As alcohol and tobacco consumption have been implicated in the pathogenesis of oral cancer, the purpose of this study was to investigate 5-HTTLPR polymorphism in patients with

Objectives: To evaluate the association between the genetic polymorphism of the tumor necrosis factor-alpha (TNF- α ) gene and the development of sepsis and septic shock in